Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Mais filtros








Intervalo de ano de publicação
2.
J Virol ; 96(13): e0071422, 2022 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-35730976

RESUMO

Pseudorabies virus (PRV) is a porcine alphaherpesvirus that belongs to the Herpesviridae family. We showed earlier that infection of porcine epithelial cells with PRV triggers activation of the nuclear factor κB (NF-κB) pathway, a pivotal signaling axis in the early immune response. However, PRV-induced NF-κB activation does not lead to NF-κB-dependent gene expression. Here, using electrophoretic mobility shift assays (EMSAs), we show that PRV does not disrupt the ability of NF-κB to interact with its κB target sites. Assessing basal cellular transcriptional activity in PRV-infected cells by quantitation of prespliced transcripts of constitutively expressed genes uncovered a broad suppression of cellular transcription by PRV, which also affects the inducible expression of NF-κB target genes. Host cell transcription inhibition was rescued when viral genome replication was blocked using phosphonoacetic acid (PAA). Remarkably, we found that host gene expression shutoff in PRV-infected cells correlated with a substantial retention of the NF-κB subunit p65, the TATA box binding protein, and RNA polymerase II-essential factors required for (NF-κB-dependent) gene transcription-in expanding PRV replication centers in the nucleus and thereby away from the host chromatin. This study reveals a potent mechanism used by the alphaherpesvirus PRV to steer the protein production capacity of infected cells to viral proteins by preventing expression of host genes, including inducible genes involved in mounting antiviral responses. IMPORTANCE Herpesviruses are highly successful pathogens that cause lifelong persistent infections of their host. Modulation of the intracellular environment of infected cells is imperative for the success of virus infections. We reported earlier that a DNA damage response in epithelial cells infected with the alphaherpesvirus pseudorabies virus (PRV) results in activation of the hallmark proinflammatory NF-κB signaling axis but, remarkably, that this activation does not lead to NF-κB-induced (proinflammatory) gene expression. Here, we report that PRV-mediated inhibition of host gene expression stretches beyond NF-κB-dependent gene expression and in fact reflects a broad inhibition of host gene transcription, which correlates with a substantial recruitment of essential host transcription factors in viral replication compartments in the nucleus, away from the host chromatin. These data uncover a potent alphaherpesvirus mechanism to interfere with production of host proteins, including proteins involved in antiviral responses.


Assuntos
Herpesvirus Suídeo 1 , Pseudorraiva , Doenças dos Suínos , Transcrição Gênica , Animais , Herpesvirus Suídeo 1/fisiologia , Interações entre Hospedeiro e Microrganismos , NF-kappa B/genética , NF-kappa B/metabolismo , Pseudorraiva/imunologia , Pseudorraiva/fisiopatologia , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/fisiopatologia
3.
J Vet Sci ; 22(2): e20, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33774936

RESUMO

BACKGROUND: Pseudorabies virus (PRV) infection leads to high mortality in swine. Despite extensive efforts, effective treatments against PRV infection are limited. Furthermore, the inflammatory response induced by PRV strain GXLB-2013 is unclear. OBJECTIVES: Our study aimed to investigate the inflammatory response induced by PRV strain GXLB-2013, establish an inflammation model to elucidate the pathogenesis of PRV infection further, and develop effective drugs against PRV infection. METHODS: Kunming mice were infected intramuscularly with medium, LPS, and different doses of PRV-GXLB-2013. Viral spread and histopathological damage to brain, spleen, and lung were determined at 7 days post-infection (dpi). Immune organ indices, levels of reactive oxygen species (ROS), nitric oxide (NO), and inflammatory cytokines, as well as levels of activity of COX-2 and iNOS were determined at 4, 7, and 14 dpi. RESULTS: At 105-106 TCID50 PRV produced obviously neurological symptoms and 100% mortality in mice. Viral antigens were detectable in kidney, heart, lung, liver, spleen, and brain. In addition, inflammatory injuries were apparent in brain, spleen, and lung of PRV-infected mice. Moreover, PRV induced increases in immune organ indices, ROS and NO levels, activity of COX-2 and iNOS, and the content of key pro-inflammatory cytokines, including interleukin (IL)-1ß, IL-6, tumor necrosis factor-α, interferon-γ and MCP-1. Among the tested doses, 10² TCID50 of PRV produced a significant inflammatory mediator increase. CONCLUSIONS: An inflammatory model induced by PRV infection was established in mice, and 10² TCID50 PRV was considered as the best concentration for the establishment of the model.


Assuntos
Herpesvirus Suídeo 1/fisiologia , Inflamação/veterinária , Pseudorraiva/imunologia , Animais , Modelos Animais de Doenças , Inflamação/imunologia , Inflamação/fisiopatologia , Inflamação/virologia , Pseudorraiva/fisiopatologia , Pseudorraiva/virologia , Sus scrofa , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/fisiopatologia , Doenças dos Suínos/virologia
4.
Vet Microbiol ; 239: 108485, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31767094

RESUMO

Pseudorabies virus (PRV) infection causes great economic losses in the pig industry. By disrupting the homeostasis of the endoplasmic reticulum (ER), many viral infections induce ER stress and trigger the unfolded protein response (UPR). However, the roles of ER stress and UPR in PRV infection remain unclear. In the present study, we demonstrate that the expression of the ER stress marker glucose-regulated protein 78 (GRP78) increased during the early stages of PRV infection, indicating that ER stress was induced. Examination of the three branches of the UPR revealed that the IRE1-XBP1 and eIF2α-ATF4 pathways were activated during PRV infection. In addition, PRV induced apoptosis in later stages of infection through the CHOP-Bcl2 axis. Overexpression of GRP78 or ER stress inducer treatment with thapsigargin could enhance PRV production. Conversely, ER stress inhibitor treatment with tauroursodeoxycholic acid reduced PRV replication. Taken together, our results reveal that PRV infection induces ER stress and activates the IRE1-XBP1 and eIF2α-ATF4 pathways.


Assuntos
Regulação da Expressão Gênica/fisiologia , Pseudorraiva/fisiopatologia , Resposta a Proteínas não Dobradas/genética , Animais , Antivirais/farmacologia , Linhagem Celular , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/fisiologia , Proteínas de Choque Térmico/genética , Transdução de Sinais/efeitos dos fármacos , Suínos , Ácido Tauroquenodesoxicólico/farmacologia , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
5.
Vet Res ; 50(1): 9, 2019 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-30717799

RESUMO

As a key cellular transcription factor that plays a central role in cellular responses to a broad range of stress factors, p53 has generally been considered as a host cell restriction factor for various viral infections. However, the defined roles of p53 in pseudorabies virus (PRV) replication, pathogenesis, and host responses remain unclear. In the present study, we initially constructed a p53 overexpressing a porcine kidney epithelial cell line (PK-15) to detect the effect of p53 on PRV replication in vitro. The results show that viral glycoprotein B (gB) gene copies and the titers of virus were significantly higher in p53 overexpressing PK-15 cells than in PK-15 and p53 inhibitor treated p53 overexpressing PK-15 cells. A similar result was also found in the p53 inhibitor PFT-α-treated PK-15 cells. We then examined the effects of p53 on PRV infection in vivo by using p53-knockout (p53-/-) mice. The results show that p53 knockout not only led to significantly reduced rates of mortality but also to reduced viral replication and development of viral encephalitis in the brains of mice following intracranial inoculation. Furthermore, we examined the effect of p53 knockout on the expression of the reported host cell regulators of PRV replication in the brains of mice by using RNA sequencing. The results show that p53 knockout downregulated the interferon (IFN) regulator genes, chemokine genes, and antiviral genes after PRV infection. This finding suggests that p53 positively regulates viral replication and pathogenesis both in vitro and in vivo. These findings offer novel targets of intrinsic host cell immunity for PRV infection.


Assuntos
Herpesvirus Suídeo 1/fisiologia , Herpesvirus Suídeo 1/patogenicidade , Imunidade Inata , Pseudorraiva/imunologia , Doenças dos Suínos/imunologia , Proteína Supressora de Tumor p53/genética , Replicação Viral , Animais , Linhagem Celular , Interações Hospedeiro-Patógeno , Pseudorraiva/fisiopatologia , Pseudorraiva/virologia , Suínos , Doenças dos Suínos/fisiopatologia , Doenças dos Suínos/virologia , Proteína Supressora de Tumor p53/metabolismo , Virulência
6.
Proc Natl Acad Sci U S A ; 115(40): E9419-E9428, 2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30154170

RESUMO

Previous studies have shown changes in membrane properties of neurons in rat deep cerebellar nuclei (DCN) as a function of development, but due to technical difficulties in obtaining viable DCN slices from adult animals, it remains unclear whether there are learning-related alterations in the membrane properties of DCN neurons in adult rats. This study was designed to record from identified DCN cells in cerebellar slices from postnatal day 25-26 (P25-26) rats that had a relatively mature sensory nervous system and were able to acquire learning as a result of tone-shock eyeblink conditioning (EBC) and to document resulting changes in electrophysiological properties. After electromyographic electrode implantation at P21 and inoculation with a fluorescent pseudorabies virus (PRV-152) at P22-23, rats received either four sessions of paired delay EBC or unpaired stimulus presentations with a tone conditioned stimulus and a shock unconditioned stimulus or sat in the training chamber without stimulus presentations. Compared with rats given unpaired stimuli or no stimulus presentations, rats given paired EBC showed an increase in conditioned responses across sessions. Whole-cell recordings of both fluorescent and nonfluorescent DCN projection neurons showed that delay EBC induced significant changes in membrane properties of evoked DCN action potentials including a reduced after-hyperpolarization amplitude and shortened latency. Similar findings were obtained in hyperpolarization-induced rebound spikes of DCN neurons. In sum, delay EBC produced significant changes in the membrane properties of juvenile rat DCN projection neurons. These learning-specific changes in DCN excitability have not previously been reported in any species or task.


Assuntos
Piscadela , Membrana Celular , Núcleos Cerebelares/fisiopatologia , Neurônios , Animais , Estimulação Elétrica , Herpesvirus Suídeo 1 , Pseudorraiva/fisiopatologia , Ratos , Ratos Long-Evans
7.
Am J Physiol Regul Integr Comp Physiol ; 314(3): R353-R365, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29118019

RESUMO

Chronic pelvic pain causes significant patient morbidity and is a challenge to clinicians. Using a murine neurogenic cystitis model that recapitulates key aspects of interstitial cystitis/bladder pain syndrome (IC), we recently showed that pseudorabies virus (PRV) induces severe pelvic allodynia in BALB/c mice relative to C57BL/6 mice. Here, we report that a quantitative trait locus (QTL) analysis of PRV-induced allodynia in F2CxB progeny identified a polymorphism on chromosome 13, rs6314295 , significantly associated with allodynia (logarithm of odds = 3.11). The nearby gene encoding acyloxyacyl hydrolase ( Aoah) was induced in the sacral spinal cord of PRV-infected mice. AOAH-deficient mice exhibited increased vesicomotor reflex in response to bladder distension, consistent with spontaneous bladder hypersensitivity, and increased pelvic allodynia in neurogenic cystitis and postbacterial chronic pain models. AOAH deficiency resulted in greater bladder pathology and tumor necrosis factor production in PRV neurogenic cystitis, markers of increased bladder mast cell activation. AOAH immunoreactivity was detectable along the bladder-brain axis, including in brain sites previously correlated with human chronic pelvic pain. Finally, AOAH-deficient mice had significantly higher levels of bladder vascular endothelial growth factor, an emerging marker of chronic pelvic pain in humans. These findings indicate that AOAH modulates pelvic pain severity, suggesting that allelic variation in Aoah influences pelvic pain in IC.


Assuntos
Hidrolases de Éster Carboxílico/metabolismo , Cistite Intersticial/enzimologia , Infecções por Escherichia coli/enzimologia , Hiperalgesia/enzimologia , Dor Pélvica/enzimologia , Pseudorraiva/enzimologia , Bexiga Urinária/inervação , Infecções Urinárias/enzimologia , Animais , Comportamento Animal , Hidrolases de Éster Carboxílico/deficiência , Hidrolases de Éster Carboxílico/genética , Cistite Intersticial/genética , Cistite Intersticial/fisiopatologia , Cistite Intersticial/psicologia , Modelos Animais de Doenças , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/fisiopatologia , Infecções por Escherichia coli/psicologia , Feminino , Predisposição Genética para Doença , Hiperalgesia/genética , Hiperalgesia/fisiopatologia , Hiperalgesia/psicologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Percepção da Dor , Limiar da Dor , Dor Pélvica/genética , Dor Pélvica/fisiopatologia , Fenótipo , Pseudorraiva/genética , Pseudorraiva/fisiopatologia , Pseudorraiva/psicologia , Locos de Características Quantitativas , Índice de Gravidade de Doença , Fator de Necrose Tumoral alfa/metabolismo , Bexiga Urinária/metabolismo , Infecções Urinárias/genética , Infecções Urinárias/fisiopatologia , Infecções Urinárias/psicologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
Virus Genes ; 53(4): 565-572, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28417300

RESUMO

Emerging pseudorabies virus (PRV) variant has led to frequent outbreaks of PRV infection among Bartha-K61-vaccinated swine population in Chinese swine farms and caused high mortality in pigs of all age since late 2011. Here, we generated a gE/gI-deleted PRV (rPRVXJ-delgI/gE-EGFP) based on PRV variant strain (PRV-XJ) through homologous DNA recombination. Compared to parental strain, rPRVXJ-delgI/gE-EGFP showed similar growth kinetics in vitro. Its safety and immunogenicity were evaluated in weaned piglets. Our results showed that piglets immunized with rPRVXJ-delgI/gE-EGFP did not exhibit any clinical symptoms, and a high level of gB-specific antibody was detected. After lethal challenge with variant PRV (PRV-FJ strain), all vaccinated piglets survived without showing any clinical symptoms except slight fever within 7 days post-challenge. In unvaccinated piglets, typical clinical symptoms of pseudorabies were observed, and the piglets were all died at 5 days post-challenge. These results indicated that a live rPRVXJ-delgI/gE-EGFP vaccine could be a maker vaccine candidate to control the currently epidemic pseudorabies in China.


Assuntos
Deleção de Genes , Herpesvirus Suídeo 1/genética , Pseudorraiva/virologia , Doenças dos Suínos/virologia , Proteínas do Envelope Viral/genética , Animais , Anticorpos Antivirais/imunologia , Herpesvirus Suídeo 1/imunologia , Herpesvirus Suídeo 1/metabolismo , Pseudorraiva/imunologia , Pseudorraiva/fisiopatologia , Pseudorraiva/prevenção & controle , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/fisiopatologia , Doenças dos Suínos/prevenção & controle , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Proteínas do Envelope Viral/administração & dosagem , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Vacinas Virais/imunologia , Desmame
9.
J Neurosci ; 32(36): 12472-87, 2012 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-22956838

RESUMO

Rabbit eyeblink conditioning is a well characterized model of associative learning. To identify specific neurons that are part of the eyeblink premotor pathway, a retrograde transsynaptic tracer (pseudorabies virus) was injected into the orbicularis oculi muscle. Four time points (3, 4, 4.5, and 5 d) were selected to identify sequential segments of the pathway and a map of labeled structures was generated. At 3 d, labeled first-order motor neurons were found in dorsolateral facial nucleus ipsilaterally. At 4 d, second-order premotor neurons were found in reticular nuclei, and sensory trigeminal, auditory, vestibular, and motor structures, including contralateral red nucleus. At 4.5 d, labeled third-order premotor neurons were found in the pons, midbrain, and cerebellum, including dorsolateral anterior interpositus nucleus and rostral fastigial nucleus. At 5 d, labeling revealed higher-order premotor structures. Labeled fourth-order Purkinje cells were found in ipsilateral cerebellar cortex in cerebellar lobule HVI and in lobule I. The former has been implicated in eyeblink conditioning and the latter in vestibular control. Labeled neurons in anterior interpositus were studied, using neurotransmitter immunoreactivity to classify individual cell types and delineate their interconnectivity. Labeled third-order premotor neurons were immunoreactive for glutamate and corresponded to large excitatory projection neurons. Labeled fourth-order premotor interneurons were immunoreactive for GABA (30%), glycine (18%), or both GABA and glycine (52%) and form a functional network within anterior interpositus involved in modulation of motor commands. These results identify a complete eyeblink premotor pathway, deep cerebellar interconnectivity, and specific neurons responsible for the generation of eyeblink responses.


Assuntos
Piscadela , Cerebelo/anatomia & histologia , Neurônios Motores/citologia , Rede Nervosa/anatomia & histologia , Músculos Oculomotores/anatomia & histologia , Pseudorraiva , Animais , Piscadela/fisiologia , Córtex Cerebelar/anatomia & histologia , Córtex Cerebelar/fisiologia , Cerebelo/fisiologia , Neurônios Motores/fisiologia , Rede Nervosa/fisiologia , Vias Neurais/anatomia & histologia , Vias Neurais/fisiologia , Músculos Oculomotores/química , Músculos Oculomotores/fisiologia , Pseudorraiva/patologia , Pseudorraiva/fisiopatologia , Coelhos
10.
Animal ; 6(11): 1821-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22717081

RESUMO

The experiment was conducted to evaluate the effects of dietary threonine (Thr) supplement on reproductive performance and immune function of the male mice challenged with pseudorabies virus (PRV). Kun-Ming male mice were assigned randomly to four groups with different Thr levels (0.70%, 0.88%, 1.10% and 1.30%). Half of the mice in each group were injected with PRV or phosphate-buffered saline (PBS) after 5 weeks' adaptation to diets. The second experiment examined the effects of dietary Thr level on copulation rate, pregnancy rate and average number per litter of PRV- or PBS-challenged male mice that copulated with adult female mice on the 9th day post PRV challenge. Sperm quality and testosterone of mice were decreased after PRV infection, but this effect was attenuated by increasing Thr levels. Copulation and conception rates were increased with increasing Thr levels (P = 0.14), but litter size was not affected (P > 0.05). In the PBS and PRV groups, mice fed higher levels of Thr had increased immunoglobulin (Ig)G, IgA and IgM concentrations. The PRV-specific antibody level, interleukin (IL)-1ß and tumor necrosis factor (TNF)-α concentration in PRV groups enhanced with increasing Thr levels; however, there was no difference in PBS groups. Furthermore, higher toll-like receptor (TLR)2 and TLR9 expressions in testis were observed by PRV challenge compared with PBS groups, and higher Thr supplement attenuated PRV-challenged induced the upregulation effect of TLR2 and TLR9 mRNA expression in testis (P < 0.05). These data suggest that higher Thr consumption was recommended in order to counteract the deleterious effects of virus invasion, possibly through the downregulated expression of TLRs, and thus to improve immunity and reproduction performance of male mice challenged with PRV.


Assuntos
Herpesvirus Suídeo 1 , Imunidade/efeitos dos fármacos , Pseudorraiva/tratamento farmacológico , Treonina/uso terapêutico , Animais , Anticorpos Antivirais/sangue , Suplementos Nutricionais , Imunoglobulina G/sangue , Interleucina-1beta/química , Masculino , Camundongos , Pseudorraiva/imunologia , Pseudorraiva/fisiopatologia , Reação em Cadeia da Polimerase em Tempo Real , Reprodução/efeitos dos fármacos , Reprodução/fisiologia , Espermatozoides/efeitos dos fármacos , Testículo/química , Testosterona/química , Testosterona/fisiologia , Treonina/administração & dosagem , Treonina/farmacologia , Fator de Necrose Tumoral alfa/química
11.
PLoS One ; 6(6): e21141, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21698154

RESUMO

Replication and transneuronal transport of pseudorabies virus (PRV) are widely used to define the organization of neural circuits in rodent brain. Here we report a dual infection approach that highlights connections to neurons that collateralize within complex networks. The method combines Cre recombinase (Cre) expression from a PRV recombinant (PRV-267) and Cre-dependent reporter gene expression from a second infecting strain of PRV (PRV-263). PRV-267 expresses both Cre and a monomeric red fluorescent protein (mRFP) fused to viral capsid protein VP26 (VP26-mRFP) that accumulates in infected cell nuclei. PRV-263 carries a Brainbow cassette and expresses a red (dTomato) reporter that fills the cytoplasm. However, in the presence of Cre, the dTomato gene is recombined from the cassette, eliminating expression of the red reporter and liberating expression of either yellow (EYFP) or cyan (mCerulean) cytoplasmic reporters. We conducted proof-of-principle experiments using a well-characterized model in which separate injection of recombinant viruses into the left and right kidneys produces infection of neurons in the renal preautonomic network. Neurons dedicated to one kidney expressed the unique reporters characteristic of PRV-263 (cytoplasmic dTomato) or PRV-267 (nuclear VP26-mRFP). Dual infected neurons expressed VP26-mRFP and the cyan or yellow cytoplasmic reporters activated by Cre-mediated recombination of the Brainbow cassette. Differential expression of cyan or yellow reporters in neurons lacking VP26-mRFP provided a unique marker of neurons synaptically connected to dual infected neurons, a synaptic relationship that cannot be distinguished using other dual infection tracing approaches. These data demonstrate Cre-enabled conditional reporter expression in polysynaptic circuits that permits the identification of collateralized neurons and their presynaptic partners.


Assuntos
Herpesvirus Suídeo 1/fisiologia , Neurônios/citologia , Pseudorraiva/fisiopatologia , Animais , Sequência de Bases , Genes Reporter , Genoma Viral , Herpesvirus Suídeo 1/genética , Masculino , Microscopia de Fluorescência , Neurônios/virologia , Fases de Leitura Aberta , Ratos , Ratos Sprague-Dawley
12.
BMC Microbiol ; 9: 246, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19948073

RESUMO

BACKGROUND: Pseudorabies virus (PRV) is an alphaherpesviruses whose native host is pig. PRV infection mainly causes signs of central nervous system disorder in young pigs, and respiratory system diseases in the adult. RESULTS: In this report, we have analyzed native host (piglets) gene expression changes in response to acute pseudorabies virus infection of the brain and lung using a printed human oligonucleotide gene set from Illumina. A total of 210 and 1130 out of 23,000 transcript probes displayed differential expression respectively in the brain and lung in piglets after PRV infection (p-value < 0.01), with most genes displaying up-regulation. Biological process and pathways analysis showed that most of the up-regulated genes are involved in cell differentiation, neurodegenerative disorders, the nervous system and immune responses in the infected brain whereas apoptosis, cell cycle control, and the mTOR signaling pathway genes were prevalent in the infected lung. Additionally, a number of differentially expressed genes were found to map in or close to quantitative trait loci for resistance/susceptibility to pseudorabies virus in piglets. CONCLUSION: This is the first comprehensive analysis of the global transcriptional response of the native host to acute alphaherpesvirus infection. The differentially regulated genes reported here are likely to be of interest for the further study and understanding of host viral gene interactions.


Assuntos
Encéfalo/metabolismo , Regulação da Expressão Gênica , Herpesvirus Suídeo 1/fisiologia , Pulmão/metabolismo , Pseudorraiva/metabolismo , Pseudorraiva/fisiopatologia , Animais , Biologia Computacional/métodos , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Suínos , Doenças dos Suínos/metabolismo , Doenças dos Suínos/fisiopatologia
13.
PLoS Pathog ; 5(10): e1000640, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19876391

RESUMO

Alpha-herpesviruses, including human herpes simplex virus 1 & 2, varicella zoster virus and the swine pseudorabies virus (PRV), infect the peripheral nervous system of their hosts. Symptoms of infection often include itching, numbness, or pain indicative of altered neurological function. To determine if there is an in vitro electrophysiological correlate to these characteristic in vivo symptoms, we infected cultured rat sympathetic neurons with well-characterized strains of PRV known to produce virulent or attenuated symptoms in animals. Whole-cell patch clamp recordings were made at various times after infection. By 8 hours of infection with virulent PRV, action potential (AP) firing rates increased substantially and were accompanied by hyperpolarized resting membrane potentials and spikelet-like events. Coincident with the increase in AP firing rate, adjacent neurons exhibited coupled firing events, first with AP-spikelets and later with near identical resting membrane potentials and AP firing. Small fusion pores between adjacent cell bodies formed early after infection as demonstrated by transfer of the low molecular weight dye, Lucifer Yellow. Later, larger pores formed as demonstrated by transfer of high molecular weight Texas red-dextran conjugates between infected cells. Further evidence for viral-induced fusion pores was obtained by infecting neurons with a viral mutant defective for glycoprotein B, a component of the viral membrane fusion complex. These infected neurons were essentially identical to mock infected neurons: no increased AP firing, no spikelet-like events, and no electrical or dye transfer. Infection with PRV Bartha, an attenuated circuit-tracing strain delayed, but did not eliminate the increased neuronal activity and coupling events. We suggest that formation of fusion pores between infected neurons results in electrical coupling and elevated firing rates, and that these processes may contribute to the altered neural function seen in PRV-infected animals.


Assuntos
Potenciais de Ação/fisiologia , Herpesvirus Suídeo 1/fisiologia , Neurônios/fisiologia , Pseudorraiva/fisiopatologia , Animais , Células Cultivadas , Eletrofisiologia , Corantes Fluorescentes/metabolismo , Células Gigantes/virologia , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , Pseudorraiva/virologia , Ratos , Ratos Sprague-Dawley , Gânglio Cervical Superior/citologia , Gânglio Cervical Superior/virologia , Suínos , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Replicação Viral
14.
PLoS Pathog ; 5(4): e1000387, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19381253

RESUMO

The neuroinvasive property of several alpha-herpesviruses underlies an uncommon infectious process that includes the establishment of life-long latent infections in sensory neurons of the peripheral nervous system. Several herpesvirus proteins are required for replication and dissemination within the nervous system, indicating that exploiting the nervous system as a niche for productive infection requires a specialized set of functions encoded by the virus. Whether initial entry into the nervous system from peripheral tissues also requires specialized viral functions is not known. Here we show that a conserved deubiquitinase domain embedded within a pseudorabies virus structural protein, pUL36, is essential for initial neural invasion, but is subsequently dispensable for transmission within and between neurons of the mammalian nervous system. These findings indicate that the deubiquitinase contributes to neurovirulence by participating in a previously unrecognized initial step in neuroinvasion.


Assuntos
Endopeptidases/fisiologia , Herpesvirus Suídeo 1/enzimologia , Pseudorraiva/virologia , Células Receptoras Sensoriais/virologia , Ubiquitina/metabolismo , Proteínas Estruturais Virais/fisiologia , Animais , Câmara Anterior/virologia , Transporte Axonal/fisiologia , Chlorocebus aethiops , Endopeptidases/genética , Infecções Oculares Virais/virologia , Herpesvirus Suídeo 1/genética , Masculino , Pseudorraiva/fisiopatologia , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Proteases Específicas de Ubiquitina , Células Vero , Proteínas Estruturais Virais/genética
15.
Virology ; 381(1): 55-66, 2008 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-18799179

RESUMO

PRV infection causes apoptosis in vitro and in vivo. However, the significance of PRV-induced apoptosis and its signaling pathways is still unknown. This work investigates the role of MAPK pathways in mediating PRV-induced apoptosis. Flow cytometry, apoptosis ELISA and western blotting using antibodies against cleaved caspase-3, -6 and PARP demonstrated that PRV induces apoptosis in a time- and dose-dependent manner. p38 and JNK/SAPK inhibitors significantly protected cells from PRV-induced apoptosis. Inhibitor treatment did not affect Us3a gene transcription and progeny virus production. Western blotting revealed that PRV activates p38 and JNK/SAPK signaling. Inhibition of NF-kappaB had no effect on PRV-mediated apoptosis. Non-replicative PRV failed to activate p38 and JNK/SAPK or induce apoptosis. PRV infection increases TNF-alpha transcription, translation and secretion, as well as TNF-alpha receptor expression. Inhibition of p38 and JNK/SAPK reduced PRV-induced TNF-alpha up-regulation. Neutralization assay confirmed that TNF-alpha is a key mediator involved in PRV-induced apoptosis.


Assuntos
Apoptose , Herpesvirus Suídeo 1/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Pseudorraiva/fisiopatologia , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Anticorpos/metabolismo , Linhagem Celular , Chlorocebus aethiops , Inibidores Enzimáticos/farmacologia , Regulação Viral da Expressão Gênica , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Transdução de Sinais/genética , Suínos , Células Vero
16.
PLoS One ; 3(5): e2096, 2008 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-18461160

RESUMO

BACKGROUND: Mast cells trigger inflammation that is associated with local pain, but the mechanisms mediating pain are unclear. Interstitial cystitis (IC) is a bladder disease that causes debilitating pelvic pain of unknown origin and without consistent inflammation, but IC symptoms correlate with elevated bladder lamina propria mast cell counts. We hypothesized that mast cells mediate pelvic pain directly and examined pain behavior using a murine model that recapitulates key aspects of IC. METHODS AND FINDINGS: Infection of mice with pseudorabies virus (PRV) induces a neurogenic cystitis associated with lamina propria mast cell accumulation dependent upon tumor necrosis factor alpha (TNF), TNF-mediated bladder barrier dysfunction, and pelvic pain behavior, but the molecular basis for pelvic pain is unknown. In this study, both PRV-induced pelvic pain and bladder pathophysiology were abrogated in mast cell-deficient mice but were restored by reconstitution with wild type bone marrow. Pelvic pain developed normally in TNF- and TNF receptor-deficient mice, while bladder pathophysiology was abrogated. Conversely, genetic or pharmacologic disruption of histamine receptor H1R or H2R attenuated pelvic pain without altering pathophysiology. CONCLUSIONS: These data demonstrate that mast cells promote cystitis pain and bladder pathophysiology through the separable actions of histamine and TNF, respectively. Therefore, pain is independent of pathology and inflammation, and histamine receptors represent direct therapeutic targets for pain in IC and other chronic pain conditions.


Assuntos
Cistite/fisiopatologia , Histamina/fisiologia , Mastócitos/fisiologia , Dor/prevenção & controle , Animais , Cistite/etiologia , Cistite/microbiologia , Herpesvirus Suídeo 1/patogenicidade , Camundongos , Dor/etiologia , Pelve/fisiopatologia , Pseudorraiva/fisiopatologia , Receptores Histamínicos/fisiologia , Doenças da Bexiga Urinária/microbiologia , Doenças da Bexiga Urinária/fisiopatologia
17.
Brain Struct Funct ; 213(1-2): 239-45, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18247051

RESUMO

Although it has been reported by several laboratories that vestibular stress activates the hypothalamo-pituitary-adrenocortical axis (HPA), the existence of neuronal connections between vestibular and hypothalamic paraventricular neurons has not yet been demonstrated. By the use of a virus-based retrograde trans-synaptic tracing technique in the rat, here we demonstrate vestibular projections to the paraventricular nucleus (PVN). Pseudorabies virus (Bartha strain, type BDR62) was injected into the PVN, and the progression of the infection along synaptically connected neurons was followed in the pons and the medulla, 3 and 4 days post-inoculation. Virus-infected neurons were revealed mainly in the medial vestibular nucleus. Labeled cells were scattered in the spinal, and very rarely in the superior nuclei, but none of them in the lateral vestibular nucleus. Injections of cholera toxin B subunit, a monosynaptic retrograde tracer into the PVN failed to label any cells in the vestibular nuclei. These results provide anatomical evidence for the existence of a vestibulo-paraventricular polysynaptic pathway and support the view that the HPA axis is modulated by vestibular stress.


Assuntos
Hipotálamo/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Estresse Fisiológico/fisiologia , Núcleos Vestibulares/metabolismo , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/metabolismo , Animais , Transporte Biológico/fisiologia , Toxina da Cólera/administração & dosagem , Toxina da Cólera/metabolismo , Herpesvirus Suídeo 1/fisiologia , Hipotálamo/patologia , Imuno-Histoquímica , Masculino , Bulbo/metabolismo , Bulbo/patologia , Bulbo/virologia , Microinjeções , Vias Neurais/metabolismo , Vias Neurais/patologia , Vias Neurais/virologia , Neurônios/metabolismo , Neurônios/patologia , Neurônios/virologia , Neurônios Eferentes/metabolismo , Neurônios Eferentes/patologia , Neurônios Eferentes/virologia , Núcleo Hipotalâmico Paraventricular/patologia , Núcleo Hipotalâmico Paraventricular/virologia , Ponte/metabolismo , Ponte/patologia , Ponte/virologia , Pseudorraiva/fisiopatologia , Pseudorraiva/virologia , Ratos , Ratos Sprague-Dawley , Medula Espinal/metabolismo , Medula Espinal/patologia , Medula Espinal/virologia , Núcleos Vestibulares/patologia , Núcleos Vestibulares/virologia
18.
Am J Physiol Regul Integr Comp Physiol ; 293(3): R1191-8, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17626130

RESUMO

Interstitial cystitis (IC) is a chronic bladder inflammatory disease of unknown etiology that is often regarded as a neurogenic cystitis. IC is associated with urothelial lesions, voiding dysfunction, and pain in the pelvic/perineal area, and diet can exacerbate IC symptoms. In this study, we used a murine neurogenic cystitis model to investigate the development of pelvic pain behavior. Neurogenic cystitis was induced by the injection of Bartha's strain of pseudorabies virus (PRV) into the abductor caudalis dorsalis tail base muscle of female C57BL/6J mice. Infectious PRV virions were isolated only from the spinal cord, confirming the centrally mediated nature of this neurogenic cystitis model. Pelvic pain was assessed using von Frey filament stimulation to the pelvic region, and mice infected with PRV developed progressive pelvic pain. Pelvic pain was alleviated by 2% lidocaine instillation into either the bladder or the colon but not following lidocaine instillation into the uterus. The bladders of PRV-infected mice showed markers of inflammation and increased vascular permeability compared with controls. In contrast, colon histology was normal and vascular permeability was unchanged, suggesting that development of pelvic pain was due only to bladder inflammation. Bladder-induced pelvic pain was also exacerbated by colonic administration of a subthreshold dose of capsaicin. These data indicate organ cross talk in pelvic pain and modulation of pain responses by visceral inputs distinct from the inflamed site. Furthermore, these data suggest a mechanism by which dietary modification benefits pelvic pain symptoms.


Assuntos
Cistite Intersticial/fisiopatologia , Dor Pélvica/fisiopatologia , Anestésicos Locais/farmacologia , Animais , Comportamento Animal/fisiologia , Capsaicina/farmacologia , Colo/fisiologia , Cistite Intersticial/etiologia , Cistite Intersticial/patologia , Azul Evans , Feminino , Herpesvirus Suídeo 1 , Lidocaína/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Dor Pélvica/etiologia , Dor Pélvica/patologia , Estimulação Física , Pseudorraiva/complicações , Pseudorraiva/fisiopatologia , Ensaio de Placa Viral
19.
J Neuroimmunol ; 180(1-2): 88-103, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16930726

RESUMO

Ba-DupGreen (BDG) is a highly attenuated, Bartha-derived pseudorabies virus (PRV) expressing green fluorescent protein (GFP) with immediate-early kinetics. Innate immune mechanisms underlying the low infectivity of the virus and the disappearance of infected neurons from the brain were studied at cellular level following injection of BDG into the spleen. The temporal shift in the expression between GFP and viral structural proteins allowed us to discriminate three stages of viral infection in the compromised neurons in correlation with the ongoing local inflammatory response. Iba1/lectin/OX42-positive microglia were recruited to infected neurons within 4-6 h following the initiation of virus replication, incorporated BrdU, isolated the infected cells before the disintegration of their membranes and phagocytosed collapsed neurons. Ex vivo-labeled blood and bone marrow-derived leukocytes, including ED-1-positive macrophages were involved in the immune cell assembly around compromised neurons, which resulted in the complete clearance of infected neurons from the early-infected brain regions.


Assuntos
Encéfalo/imunologia , Encéfalo/virologia , Herpesvirus Suídeo 1/imunologia , Imunidade Inata/imunologia , Pseudorraiva/imunologia , Animais , Transporte Axonal/imunologia , Morte Celular/imunologia , Linhagem Celular Transformada , Quimiotaxia de Leucócito/imunologia , Modelos Animais de Doenças , Progressão da Doença , Gliose/imunologia , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/patogenicidade , Macrófagos/imunologia , Masculino , Microglia/imunologia , Vias Neurais/imunologia , Vias Neurais/virologia , Neurônios/imunologia , Neurônios/virologia , Fagocitose/imunologia , Pseudorraiva/fisiopatologia , Ratos , Ratos Wistar , Sus scrofa , Fatores de Tempo , Replicação Viral/imunologia
20.
J Virol ; 80(11): 5571-6, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16699038

RESUMO

Neurotropism is a distinctive feature of members of the Alphaherpesvirinae. However, its molecular basis remains enigmatic. In the past, research has been focused mainly on the role of viral envelope proteins in modulating herpesvirus neuroinvasion and neurovirulence (T. C. Mettenleiter, Virus Res. 92:192-206, 2003). To further analyze the molecular requirements for neuroinvasion of the alphaherpesvirus pseudorabies virus (PrV), adult mice were infected intranasally with a set of single- or multiple-deletion mutants lacking the UL3, UL4, UL7, UL11, UL13, UL16, UL17, UL21, UL31, UL34, UL37, UL41, UL43, UL46, UL47, UL48, UL51, US3, US9, glycoprotein E (gE), gM, UL11/US9, UL11/UL16, UL16/UL21, UL11/UL16/UL21, UL11/gE, UL11/gM, UL43/gK, UL43/gM, or UL43/gK/gM genes. Neurovirulence was evaluated by measuring mean survival times compared to that after wild-type virus infection. Furthermore, by immunohistochemical detection of infected neurons, the kinetics of viral spread in the murine central nervous system was investigated.


Assuntos
Herpesvirus Suídeo 1/química , Neurônios/virologia , Pseudorraiva/fisiopatologia , Proteínas do Envelope Viral/fisiologia , Animais , Antígenos Virais/imunologia , Viroses do Sistema Nervoso Central/virologia , Cinética , Camundongos , Pseudorraiva/virologia , Proteínas do Envelope Viral/genética , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA